Language selection

Search

Patent 2807418 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2807418
(54) English Title: SIMPLIFIED BASIC MEDIA FOR HUMAN PLURIPOTENT CELL CULTURE
(54) French Title: MILIEUX BASIQUES SIMPLIFIES DESTINES A LA CULTURE DE CELLULES PLURIPOTENTES HUMAINES
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12N 5/00 (2006.01)
(72) Inventors :
  • THOMSON, JAMES A. (United States of America)
  • CHEN, GUOKAI (United States of America)
(73) Owners :
  • WISCONSIN ALUMNI RESEARCH FOUNDATION (United States of America)
(71) Applicants :
  • WISCONSIN ALUMNI RESEARCH FOUNDATION (United States of America)
(74) Agent: GOWLING WLG (CANADA) LLP
(74) Associate agent:
(45) Issued: 2017-04-04
(86) PCT Filing Date: 2011-08-05
(87) Open to Public Inspection: 2012-02-09
Examination requested: 2013-02-21
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2011/046796
(87) International Publication Number: WO2012/019122
(85) National Entry: 2013-02-01

(30) Application Priority Data:
Application No. Country/Territory Date
61/371,128 United States of America 2010-08-05

Abstracts

English Abstract

Fully defined media that support pluripotent cell viability, proliferation, cloning, and derivation, as well as methods and compositions including these media are described. Methods for deriving iPS cells from adult individuals under defined, xeno-free conditions are also described.


French Abstract

La présente invention concerne des milieux entièrement définis aidant à la viabilité, la prolifération, le clonage, et la dérivation de cellules pluripotentes, ainsi que des procédés et des compositions comprenant ces milieux. L'invention concerne également des procédés de dérivation de cellules souches pluripotentes induites (iPS) à partir d'individus adultes dans des conditions définies, exemptes de substances étrangères.

Claims

Note: Claims are shown in the official language in which they were submitted.


WHAT IS CLAIMED IS:
1. A defined, albumin-free medium free of any component obtained from a non-
human
animal, the medium comprising water, salts, amino acids, vitamins, a carbon
source,
insulin, FGF2, selenium, transferrin, L-ascorbic acid, and one of TGF-.beta.
and NODAL,
each combined in an amount that supports proliferation of pluripotent stem
cells.
2. A method for culturing pluripotent stem cells, the method comprising the
steps of:
placing pluripotent stem cells on a matrix; and
contacting the cells with the defined, albumin-free medium of Claim 1.
3. The method of Claim 2, wherein the matrix comprises laminin.
4. The method of Claim 2, wherein the matrix comprises vitronectin.
5. The method of Claim 2, wherein the cells are contacted with the medium
under hypoxic
conditions.
6. The method of Claim 2, wherein the pluripotent stem cells are embryonic
stem cells.
7. The method of Claim 2, wherein the pluripotent stem cells are induced
pluripotent stem
cells.
8. The method of Claim 2, wherein the pluripotent stem cells are human
cells.
9. A method for cloning a pluripotent stem cell, the method comprising the
step of:
plating pluripotent stem cells at cloning density in a defined, albumin-free
medium that is free of any component obtained from a non-human animal, the
medium comprising the following components: water, salts, amino acids,
vitamins, a carbon source, insulin, FGF2, selenium, transferrin, L-ascorbic
acid,
and one of TGF-.beta. and NODAL, each combined in an amount that supports
pluripotent stem cell cloning.

- 24 -

10. The method of Claim 9, wherein the medium further comprises a ROCK
inhibitor.
11. The method of Claim 10, wherein the ROCK inhibitor is selected from the
group
consisting of HA100 and Y27632.
12. The method of Claim 11, wherein the medium further comprises
blebbistatin.
13. A method of cryopreserving pluripotent stem cells, the method
comprising the step of:
freezing pluripotent stem cells in a defined, albumin-free medium free of any
component obtained from a non-human animal, the medium comprising the
following components: water, salts, amino acids, vitamins, a carbon source,
insulin, FGF2, selenium, transferrin, L-ascorbic acid, and one of TGF-.beta.
and
NODAL.
14. A method for deriving an induced pluripotent stem (iPS) cell under
defined conditions,
the method comprising the step of:
reprogramming a somatic cell in a defined, albumin-free medium free of any
component obtained from a non-human animal, the medium comprising the
following components: water, salts, amino acids, vitamins, a carbon source,
insulin, FGF2, selenium, transferrin, L-ascorbic acid, and one of TGF-.beta.
and
NODAL, each combined in an amount that supports somatic cell reprogramming
to derive an iPS cell.
15. The method of Claim 14, wherein the reprogramming step comprises
contacting the cell
with TGF-.beta. for 5-10 days.
16. The method of Claim 15, further comprising the steps of:
removing the TGF-.beta. after 5-10 days; and
contacting the cells with a medium consisting essentially of:
water, salts, amino acids, vitamins, a carbon source, insulin, an FGF,
selenium, L-
ascorbic acid, and transferrin.
- 25 -

17. The method of Claim 14, wherein the reprogramming step comprises
contacting the cell
with hydrocortisone.
18. The method of Claim 14, wherein the reprogramming step comprises
contacting the cell
with butyrate.
19. The method of Claim 14, wherein the iPS cell is derived under xeno-free
conditions.
20. The method of Claim 14, wherein the somatic cell is an adult somatic
cell.
21. The method of Claim 14, wherein the reprogramming step comprises using
a viral vector.
22. The method of Claim 14, wherein the reprogramming step comprises using
an episomal
vector.
23. A defined, albumin-free growth medium that supports long-term culture
of pluripotent
stem cells, wherein the medium is free of any component obtained from a non-
human
animal and consists essentially of the following components:
water, salts, amino acids, vitamins, a carbon source, insulin, an FGF,
selenium,
transferrin, L-ascorbic acid, and one of TGF-.beta. and NODAL, each combined
in an
amount sufficient to support long-term pluripotent stem cell culture.
24. A method for culturing pluripotent stem cells, the method comprising
the steps of:
placing pluripotent stem cells on a matrix; and
contacting the cells with a defined, albumin-free medium that is free of any
component obtained from a non-human animal and consists essentially of the
following components:
water, salts, amino acids, vitamins, a carbon source, insulin, an FGF,
selenium,
transferrin, L-ascorbic acid, and one of TGF-.beta. and NODAL, each combined
in an
amount sufficient to support long-term pluripotent stem cell culture.
- 26 -

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 02807418 2015-03-25
SIMPLIFIED BASIC MEDIA FOR HUMAN PLURIPOTENT CELL CULTURE
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] Not applicable.
STATEMENT REGARDING FEDERALLY SPONSORED
RESEARCH OR DEVELOPMENT
[0002] This invention was made with government support under ES017166
awarded by
the National Institutes of Health. The government has certain rights in the
invention.
BACKGROUND
[0003] Pluripotent cells, such as embryonic stem (ES) cells and induced
pluripotent stem
(iPS) cells, have the potential to differentiate into cells of all three
primary germ layers
(Thomson, etal., Science 282, 1145-1147 (1998)). The remarkable developmental
potential of
pluripotent cells has proven useful for basic research and clinical
application. Many basic
methods for human pluripotent cell culture, such as growth media, plate
coating, and other
conditions, have been developed and refined (Ludwig etal., Nat. Biotechnol 24,
185-187 (2006);
Ludwig et al., Nat. Methods 3, 637-646 (2006)). For example, while human ES
cells were
initially cultured in fetal bovine serum (FBS)-containing media on murine
embryonic fibroblast
(MEF) feeder cells, fully defined media as well as defined protein matrices
are now available
(Ludwig et al., Nat. Biotechnol 24, 185-187 (2006)).
[0004] Over the past ten years, pluripotent cell culture methods have
evolved
considerably. Several growth media were developed that provide basic nutrients
and growth
factors for survival and expansion of pluripotent cells and directly determine
how cells grow and
differentiate. TeSRTm was one of the first defined media that supports
pluripotent cell
maintenance in an undifferentiated state in the absence of feeder cells or
conditioned medium
through multiple culture passages (Ludwig et al., Nat. Methods 3, 637-646
(2006); US Patent
No. 7,449,334). TeSRTm contains 18 components in addition to the basal medium
DMEM/F12
that itself has 52 components (Table 1).
-1-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[0005] The variety of different growth media available for pluripotent
cell culture
contributes to inconsistencies in research findings. The media that are
presently used for
pluripotent cell derivation and growth, including fully defined media, contain
components that
can influence pluripotent cells in various ways. Prior to the invention
described herein, it was
not known how each media component, alone or in combination with other
components, affects
various pluripotent cell functions such as viability, pluripotency, or
differentiation in cell culture.
[0006] For example, albumin, the most abundant protein component present
in most
media, is a lipid carrier and, as such, can affect differentiation or
maintenance of pluripotency via
its associated lipids. The qualities of albumin and of its associated lipids
determine whether it
can be used for human pluripotent cell culture. However, albumin quality
varies greatly
depending on its source, even when produced from a recombinant genetic
material, contributing
to variations between experiments conducted under otherwise equivalent
conditions. Also, while
cloned human serum albumin is available, it is seldom used for routine
experimentation due to its
comparatively high cost.
[0007] Efforts to eliminate albumin from the medium have proved
unsuccessful.
Omission of albumin, or any other growth factor present in TeSR, led to a
dramatic decline in
human ESC culture performance, such as decreased cell viability,
proliferation, and pluripotency
(Ludwig et at., Nat. Biotechnol 24, 185-187 (2006)).
[0008] To fully exploit the potential of pluripotent cells for drug
discovery, testing, and
transplantation therapy, derivation and growth of these cells under fully-
defined and, ideally,
xeno-free, conditions is desirable. There is, thus an unmet need in the art
for media free of
components that introduce inconsistencies to maintain control over pluripotent
cell culture
conditions. Specifically, there is a need in the art for pluripotent cell
culture media containing
only those components that support pluripotent cell functions important for a
specific culture
objective.
BRIEF SUMMARY
[0009] The invention relates generally to media, compositions, and
methods for deriving
and culturing pluripotent cells, and more particularly, to fully-defined media
for pluripotent cells.
[00010] In a first aspect, the present invention is summarized as albumin-
free media that
support viability, growth, and pluripotency of pluripotent cells.
-2-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00011] In some embodiments of the first aspect, the medium contains
selenium.
[00012] In some embodiments of the first aspect, the medium contains
NODAL.
[00013] In some embodiments of the first aspect, the medium contains
transferrin.
[00014] In some embodiments of the first aspect, the medium contains
transforming
growth factor beta (TGF-I3).
[00015] In some embodiments of the first aspect, the medium contains only
water, salts,
amino acids, vitamins, a carbon source, insulin, and a fibroblast growth
factor (FGF), each in
amounts sufficient to support pluripotent stem cell viability.
[00016] In some embodiments of the first aspect, the medium contains only
water, salts,
amino acids, vitamins, a carbon source, insulin, an FGF, selenium,
transferrin, and one of TGF-I3
and NODAL, each in an amount sufficient to support pluripotent stem cell
proliferation.
[00017] In some embodiments of the first aspect, the medium supports
survival after
passaging, freezing, proliferation, pluripotency, derivation, and cloning of
pluripotent cells.
[00018] In some embodiments of the first aspect, the medium is xeno-free.
[00019] In a second aspect, the present invention is summarized as a
method for culturing
pluripotent stem cells in a defined medium. In some embodiments of the second
aspect, the
medium used to culture pluripotent cells contains only water, salts, amino
acids, vitamins, a
carbon source, insulin, and an FGF, each in amounts sufficient to support
pluripotent cell
viability. In some embodiments of the second aspect, the medium used to
culture pluripotent
cells contains only water, salts, amino acids, vitamins, a carbon source,
insulin, an FGF,
selenium, transferrin, and one of TGF-13 and NODAL, each in an amount
sufficient to support
pluripotent stem cell proliferation. In some embodiments of the second aspect,
the medium
contains defined factors that support extended growth, pluripotency, cloning,
freezing, or
derivation of pluripotent cells. In some embodiments of the second aspect, the
medium used to
culture pluripotent cells is xeno-free.
[00020] In a third aspect, the present invention is directed to an in
vitro cell culture
composition of pluripotent cells in a medium that is substantially free of 13-
mercaptoethanol and
albumin. In some embodiments of the third aspect, the culture composition is
free of fibroblast
feeder cells, conditioned medium, and xeno-contamination.
[00021] In a fourth aspect, the present invention is summarized as a
method for deriving
iPS cells from an adult individual under completely defined conditions. The
method includes the
-3-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
steps of culturing a somatic cell from an adult individual in a medium
containing water, salts,
amino acids, vitamins, a carbon source, insulin, and an FGF, all in sufficient
amount to maintain
viability, and reprogramming the cell in defined conditions such as to derive
iPS cells.
[00022] In some embodiments of the fourth aspect, the medium contains TGF-
I3 during
parts or all of the reprogramming process.
[00023] In some embodiments of the fourth aspect, the medium contains
butyrate.
[00024] In some embodiments of the fourth aspect, the medium contains
hydrocortisone.
[00025] In some embodiments of the fourth aspect, the medium is xeno-free.
[00026] In a fifth aspect, the present invention is summarized as a method
for cloning a
pluripotent stem cell in an albumin-free medium. The method includes the step
of plating
pluripotent stem cells at cloning density in an albumin-free medium that
supports pluripotent
stem cell cloning.
[00027] In some embodiments of the fifth aspect, the medium contains a
ROCK inhibitor.
[00028] In some embodiments of the fifth aspect, the medium contains
blebbistatin.
[00029] In some embodiments of the fifth aspect, the medium contains only
water, salts,
amino acids, vitamins, a carbon source, insulin, an FGF, selenium,
transferrin, and one of TGF-I3
and NODAL, each in an amount sufficient to support pluripotent stem cell
cloning.
[00030] In a sixth aspect, the present invention is summarized as a method
of
cryopreserving pluripotent stem cells in an albumin-free medium. The method
includes the step
of freezing pluripotent stem cells in an albumin-free medium.
[00031] In some embodiments of the sixth aspect, the medium contains only
water, salts,
amino acids, vitamins, a carbon source, insulin, an FGF, selenium,
transferrin, one of TGF-I3 and
NODAL, and dimethyl sulfoxide (DMSO).
[00032] In a seventh aspect, the invention is summarized as an iPS cell
derived under
albumin-free conditions. iPS cells derived in the absence of albumin are free
of endogenous
albumin contaminations.
[00033] The methods and compositions described herein are useful in a
variety of
applications for deriving, culturing, and using pluripotent cells. It is an
object of the present
invention to define short term and long term culture conditions for
pluripotent cells limited to
factors that support the intended culture objective.
-4-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00034] It is another object of the present invention to provide culture
conditions for
pluripotent cells that maximize percentage of cultured cells in an
undifferentiated state.
[00035] It is another object of the present invention to provide media
that can serve as the
platform necessary to examine how various conditions affect pluripotent cells
and to compare
experiments previously reported in different media backgrounds.
[00036] These and other features, objects, and advantages of the present
invention will
become better understood from the description that follows. In the
description, reference is made
to the accompanying drawings, which form a part hereof and in which there is
shown by way of
illustration, not limitation, embodiments of the invention. The description of
preferred
embodiments is not intended to limit the invention to cover all modifications,
equivalents and
alternatives. Reference should therefore be made to the claims recited herein
for interpreting the
scope of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[00037] The present invention will be better understood and features,
aspects and
advantages other than those set forth above will become apparent when
consideration is given to
the following detailed description thereof. Such detailed description makes
reference to the
following drawings, wherein:
[00038] FIG. 1 A-E illustrate media elements for human ES cell survival
and self-renewal
in culture. FIG lA illustrates 24 hour survival indices for individualized
cells plated in the
various media. Media abbreviations are as listed in Table 1. The presence of
insulin and
fibroblast growth factor (IF), bovine serum albumin (BSA), beta-mecaptoethanol
(BME) is
indicated by "+" and absence is indicated by "-." FIG. 1B illustrates 24 hour
or 96 hour survival
indices for individualized cells plated in the various media. The addition of
insulin and
fibroblast growth factor (FGF) is indicated by "+" and removal is indicated by
"-." FIG. 1C
illustrates 24 hour or 129 hour survival indices for individualized cells
cultured in TeSRTm
medium with Vitamin C (TeSR), TeSRTm medium without Vitamin C (TeSRTm-LAA), or
DF5
medium. FIG. 1D illustrates cell proliferation after each of three passages in
DF5, DF5 with
added Selenium (DF5+Selenium), DF12, or DF12 from which Selenium had been
removed
(DF12-Selenium). FIG. lE illustrates a comparative analysis of twelve
different base media.
-5-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00039] FIG. 2 A-F illustrate optimization of human ES cell and iPS cell
culture
conditions with DF5S. FIG. 2A shows survival indices for individualized cells
that were seeded
at low density (-1,500cells/cm2) in either DF5S (bottom) or TeSRTm (top) and
cultured at
different 02 and CO2 concentrations (015C5: 15% 02 and 5% CO2; 015C10: 15% 02
and 10%
CO2; 05C10: 5% 02 and 10% CO2). Cell survival was examined at 24 hours and 124
hours.
FIG. 2B shows the cloning efficiency of H1 cells cultured in various media in
the presence
(+HA100) or absence of the small molecule HA100 (CM100: conditioned media with
100ng/m1
FGF). FIG. 2C shows the cloning efficiency of H1 cells and iPS cells derived
from foreskin
fibroblasts in various media. FIG. 2D shows the cloning efficiency of iPS
cells derived from
foreskin fibroblasts in various media. DF5S trFe indicates DF5S media to which
holotransferrin
was added. FIG. 2E illustrates the cloning efficiency of H1 cells cultured in
various media in the
presence of HA100 (10 M, 24 hours), blebbistatin (10 M, 4 hours), or Y27632
(10 M, 24
hours), compared to cloning efficiency in the absence of these factors
(control). Asterisks
indicate p<0.05. FIG. 2F illustrates cloning efficiency of H1 cells in
conditioned medium (CM),
CM with ROCK inhibitor (HA100), TeSR with ROCK inhibitor, and E8 with ROCK
inhibitor in
normoxic (dark gray bars) or hypoxic (light gray bars) conditions. Error bars
indicate the
standard error of the mean; asterisks indicate p<0.05.
[00040] FIG. 3 A-B illustrate pluripotent cell growth and gene expression
in DMEM/F12
supplemented with insulin, transferrin, selenium, L-ascorbic acid, FGF2, and
TGF- 0 or NODAL
(referred to herein as "E8 (TGF- 13)" and "E8 (NODAL)," respectively). FIG. 3A
illustrates fold
expansion of H1 ES cells (top) and iPS cells (bottom) maintained in TeSRTm
(dark gray lines) or
E8 (TGF- 13) (light gray lines). FIG. 3B illustrates global gene expression of
H1 ES cells grown
in E8 (TGF- 13) and H1 ES cells grown in TeSRTm. RNA of H1 cells maintained in
either TeSR
or E8 (TGF13) medium for 3 passages was analyzed by RNA-seq with Illumina
Genome
Analyzer GAIIX (global gene expression correlation R = 0.954 (Spearman
Correlation)).
[00041] FIG. 4A-F illustrate iPS cell derivation under defined conditions.
FIG. 4A shows
proliferation of foreskin fibroblasts in DF5SFe-based media to which various
fibroblast growth
factors (FGF) were added, compared to proliferation in FBS-containing media.
FIG. 4B shows
fibroblast growth in various media supplemented with hydrocortisone. FIG. 4C
shows
expression of the pluripotency markers OCT4 (left) and SSEA4 (right). FIG. 4D
illustrates
expression of selected genes by foreskin fibroblasts, hES cells, iPS cells
derived on feeder cells
-6-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
(iPS Cells (Feeder)), and iPS cells derived in E8 medium (iPS Cells (E8)). All
cells were
maintained in E8 (TGFI3) medium prior to RNA analysis, except for fibroblasts,
which were
maintained in E8 with hydrocortisone. FIG. 4E illustrates global gene
expression of human ES
and iPS cells derived in E8 (TGFI3) media (R = 0.955). FIG. 4F global gene
expression of iPS
cells derived on MEF and iPS cells derived in E8 (TGFI3) media.
[00042] FIG. 5A-C illustrate media improvement for iPS cell derivation.
FIG 5A shows
proliferation of foreskin (dark grey bars) and PRPF8-2 adult fibroblasts
(light grey bars) in
DF5SFe media supplemented with TGF-I3, hydrocortisone, TGF-I3 and
hydrocortisone, or TGF-I3
and hydrocortisone without FGF. FIG. 5B illustrates the effect of TGF-I3 and
butyrate on
reprogramming of foreskin fibroblasts. Four to five weeks after initial
reprogramming
transfection, colony numbers for transformed cells and true iPS cells were
scored and the ratio of
iPS colonies to non-iPS cell colonies was calculated.
[00043] FIG. 6A-B illustrate derivation of iPS cells from adult
fibroblasts under fully-
defined conditions without secondary passage. FIG. 6A illustrates an example
of a
reprogramming protocol. FIG 6B illustrates expression of the pluripotency
markers OCT4 and
SSEA4, as determined by flow cytometric analysis of iPSC lines maintained in
DMEM/F12
supplemented with insulin, transferrin, selenium, L-ascorbic acid, FGF2, and
TGF- 0 or NODAL
("E8") for 20 passages. Shaded peak: staining with antibodies specific to OCT4
(left) and
SSEA4 (right); unshaded peak: mouse IgG control antibody.
[00044] FIG. 7A-C illustrate reprogramming efficiency of human fibroblasts
in various
media. FIG. 7A illustrates the number of iPS cell colonies per 80,000
fibroblasts subjected to
reprogramming with mouse fibroblast feeder cells (MEF) or in E8-based medium.
To improve
efficiency, 100 ILIM sodium butyrate was added to both conditions. FIG. 7B
illustrates the
number of iPS cell colonies per 80,000 fibroblasts subjected to reprogramming
in TeSRTm or in
E8-based medium. FIG. 7C illustrates the effects of TGF-I3 and butyrate
exposure time on
reprogramming efficiency of foreskin fibroblasts under fully-defined
conditions. Fibroblasts
were reprogrammed in DMEM/F12 supplemented with insulin, transferrin,
selenium, L-ascorbic
acid, and FGF2 (E8 without TGF- 0) or in E8, in the presence or absence of 100
M butyrate.
Reprogramming efficiency for all conditions was analyzed after 30 days after
reprogramming.
Asterisks indicate p<0.05.
-7-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00045] While the present invention is susceptible to various
modifications and alternative
forms, exemplary embodiments thereof are shown by way of example in the
drawings and are
herein described in detail. It should be understood, however, that the
description of exemplary
embodiments is not intended to limit the invention to the particular forms
disclosed, but on the
contrary, the intention is to cover all modifications, equivalents and
alternatives falling within
the spirit and scope of the invention as defined by the appended claims.
DESCRIPTION OF EXEMPLARY EMBODIMENTS
[00046] The present invention relates to the inventors' observation that
certain media
components once thought to be essential to culturing pluripotent cells can be
omitted from
pluripotent cell culture media formulated to achieve certain culture
objectives.
[00047] As used herein, the term "pluripotent cell" means a cell capable
of differentiating
into cells of all three germ layers. Examples of pluripotent cells include
embryonic stem cells
and induced pluripotent stem (iPS) cells. As used herein, "iPS cells" refer to
cells that are
substantially genetically identical to their respective differentiated somatic
cell of origin and
display characteristics similar to higher potency cells, such as ES cells, as
described herein. The
cells can be obtained by reprogramming non-pluripotent (e.g. multipotent or
somatic) cells.
[00048] The invention relates to new media free of factors not essential
for a particular
culture objective. Examples of culture objectives include, but are not limited
to, cell survival,
passaging, proliferation, pluripotency, cloning, and iPS cell derivation.
Specifically, the
invention relates to albumin-free media.
[00049] As a point of clarification, "passaging" and "cloning" are
distinct methods.
"Passaging" describes the process of dividing cells that have been cultivated
in a culture vessel
up to a certain density into aggregates, which are then placed into new
culture vessels. These
aggregates can contain any number of cells, typically between 100 to 1,000
cells, which readily
initiate growth in culture. In contrast, "cloning" refers to initiating clonal
colonies by growing
human ES cell colonies from single individual ES cells. As used herein,
"cloning efficiency"
means the number of individualized cells that form new cell colonies divided
by the number of
individualized cells plated in culture. Cloning efficiency varies considerably
depending on
culture conditions. For example, the cloning efficiency of human ES cells
under defined and
xeno-free conditions on MATRIGEL is very low (i.e., less than about 0.1%),
while cloning
-8-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
efficiency of these cells cultured with fibroblast-conditioned medium, while
still low (i.e., less
than about 2%), is high enough to initiate clonal ES cell colonies.
[00050] Certain media components presently used can be damaging to the
cultured cells or
induce differentiation. For example, 13-mercaptoethanol can damage and even
kill cultured
pluripotent cells. Serum media additives, such as bovine serum albumin (BSA)
or fetal calf
serum (FCS), can induce differentiation of cultured pluripotent cells. Also,
commercially
available serum components can differ significantly in their composition, even
when supplied
from the same source, introducing unpredictable culture variability. The media
described herein
are substantially free of damaging, differentiating, and undefined factors
present in most
conventional pluripotent cell culture media. The disclosed media have been
successfully used
for various culture objectives, such as supporting short term pluripotent
cells viability, e.g., 24
hrs, short term proliferation, e.g., 4-5 days, maintaining pluripotent cells
for extended culture
periods, e.g., more than 25 passages in 3 months, and to derive iPS cells from
both fetal and
adult fibroblasts with lentiviral and episomal vectors.
[00051] New minimal media specifically tailored for certain cell culture
objectives were
developed. Various media components, such as salts, vitamins, glucose sources,
minerals, and
amino acids were tested, alone or in combination, to determine their
individual effect on
viability, proliferation, or pluripotency. A new survival assay was developed
and used to
determine which components are essential for pluripotent cell survival after
dissociation. New
media were tested for their ability to support proliferation and sustain
pluripotency. These media
were also used in cloning assays to determine how each medium affects single
cells and their
cloning efficiency. A complete list of ingredients for each new medium
described herein is set
forth in Table 1 (light and dark shaded fields indicate presence of a
component in the medium,
checkered fields indicate interchangeable components, clear fields indicate
absence of a
component in the medium).
-9-

CA 02807418 2013-02-01
WO 2012/019122
PCT/US2011/046796
[00052] Table 1. Media compositions.
Components GF
NM DF5 DF5S DF5SFe DF12 TeSR DF5S+ DF5S+ DF5S+ E8
TGF-B NODAL HCort
DMEM/F 12 =i1i1i1i1i1i1i,
1111111111111111111111111111111111111111111111
NaHCO3 =11111111111N
1111111111111111111111111111111111111111111111
1111111111111111111111111111111111111111111111111111111111111111111111111111111
11M11111
L-Ascorbic Acid
Glutathione
L-Glutamine
Defined lipids
Thiamine
Trace elements B
Trace elements C
BME
BSA
Insulin
FGF2
TGF-I3 1111111111111111111111111111
i ii
Pipecolic acid !!!!!!!!!!!!!!!!!!!!!!!!!!!!
LiC1
GABA
H20
Nodal
...........
Hydrocortisone
[00053] The various media described herein can be prepared from the basic
ingredients.
Alternatively, one of skill in the art appreciates the advantageous efficiency
of using a basal
media as starting material to prepare the disclosed new media. The term "basal
medium" as used
herein means a medium that supports growth of certain single-celled organisms
and cells that do
not require special media additives. Typical basal medium components are known
in the art and
include salts, amino acids, vitamins, and a carbon source (e.g., glucose).
Other components that
do not change the basic characteristic of the medium but are otherwise
desirable can also be
included, such as the pH indicator phenol red. For example, Dulbecco's
Modified Eagle
Medium: Nutrient Mixture F-12 (DMEM/F12) is a basal medium commonly used to
make
suitable growth media for mammalian cell culture. A complete list of
ingredients of DMEM/F12
is set forth in Table 2.
-10-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00054] Table 2. DMEM : F-12 Medium Formulation (ATCC Catalog No. 30-
2006).
Inorganic Salts (g/liter) Amino Acids (g/liter) Vitamins
(g/liter) Other (g/liter)
CaC12 (anhydrous) L-Alanine 0.00445 D-Biotin 0.00000365 D-Glucose
3.15100
0.11665 L-Arginine=HC10.14750 Choline Chloride 0.00898 HEPES
3.57480
CuSO4 (anhydrous) L-Asparagine=H20 Folic Acid 0.00265 Hypoxanthine
0.00239
0.0000008 0.00750 myo-Inositol 0.01261 Linoleic Acid
0.000044
Fe(NO3)3.9H20 0.00005 L-Aspartic Acid 0.00665 Niacinamide 0.00202 Phenol
Red, Sodium Salt
FeSO4.7H20 0.000417 L-Cystine=HC1=H20 D-Pantothenic Acid
0.00810
Mg504 (anhydrous) 0.01756 0.00224
Putrescine=2HC10.00008
0.08495 L-Cystine=2HC10.03129 Pyridoxine=HC10.00203
Pyruvic Acid=Na 0.05500
KC10.3118 L-Glutamic Acid 0.00735 Riboflavin 0.00022 DL-Thioctic
Acid
NaHCO3 1.20000 L-Glutamine 0.36510 Thiamine=HC10.00217 0.000105
NaC17.00000 Glycine 0.01875 Vitamin B-12 0.00068 Thymidine
0.000365
Na2HPO4 (anhydrous) L-Histidine=HC1=H20
0.07100 0.03148
NaH2PO4 H2O. 0.06250 L-Isoleucine 0.05437
Zn504=7H20 0.000432 L-Leucine 0.05895
L-Lysine=HC10.09135
L-Methionine 0.01724
L-Phenylalanine 0.03548
L-Proline 0.01725
L-Serine 0.02625
L-Threonine 0.05355
L-Tryptophan 0.00902
L-Tyrosine=2Na=2H20
0.05582
L-Valine 0.05285
[00055] Unless defined otherwise, all technical and scientific terms used
herein have the
same meaning as commonly understood by one of ordinary skill in the art to
which the invention
pertains. Although any methods and materials similar to or equivalent to those
described herein
can be used in the practice or testing of the present invention, the preferred
methods and
materials are described herein.
[00056] In describing the embodiments and claiming the invention, the
following
terminology will be used in accordance with the definitions set out below.
[00057] As used herein, "about" means within 5% of a stated concentration
range or
within 5% of a stated time frame.
[00058] As used herein, "essentially serum-free" means that a medium does
not contain
serum or serum replacement, or that it contains essentially no serum or serum
replacement. For
example, an essentially serum-free medium can contain less than about 10%, 9%,
8%, 7%, 6%,
5%, 4%, 3%, 2% or 1% serum, wherein the culturing capacity of the medium is
still observed.
[00059] The term "defined culture medium" or "defined medium," as used
herein, means
that the identity and quantity of each medium ingredient is known.
-11-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00060] As used herein, "a medium consisting essentially of' means a
medium that
contains the specified ingredients and, optionally, other ingredients that do
not materially affect
its basic characteristics.
[00061] As used herein, "effective amount" means an amount of an agent
sufficient to
evoke a specified cellular effect according to the present invention.
[00062] As used herein, "viability" means the state of being viable.
Pluripotent cells that
are viable attach to the cell plate surface and do not stain with the dye
propidium iodide absent
membrane disruption. Short term viability relates to the first 24 hours after
plating the cells in
culture. Typically, the cells do not proliferate in that time.
[00063] As used herein, "short term growth" means cell proliferation for 4-
5 days in
culture.
[00064] As used herein, "extended growth" means growth for at least five
passages.
Typically, media are tested for their ability to support pluripotent cell
growth for more than
twenty passages (approximately 2-3 months).
[00065] As used herein, "long-term culture" means more than 15 passages
(approximately
two months in culture).
[00066] As used herein, "pluripotency" means a cell's ability to
differentiate into cells of
all three germ layers.
[00067] As used herein, "cloning" means a process of initiating a cell
culture from a
starting culture, ideally, from a single pluripotent cell or at least from
very few cells. Culture
conditions that permit clonal culture of undifferentiated pluripotent cells
may be the most
demanding conditions of all of those required in normal pluripotent cell
culture and proliferation.
[00068] As used herein, "iPS cell derivation" means reprogramming a cell
that is not
pluripotent to become pluripotent.
[00069] As used herein, "xeno-free" means cell culture conditions free of
any cell or cell
product of species other than that of the cultured cell.
[00070] As used herein, "normoxic condition" means conditions with about
20% oxygen.
[00071] As used herein, "hypoxic condition" means conditions with less
than about 20%
oxygen, e.g., about 5% oxygen.
[00072] The invention will be more fully understood upon consideration of
the following
non-limiting Examples.
-12-

CA 02807418 2015-03-25
EXAMPLES
[00073] Example 1: Pluripotent cell survival assay.
[00074] Five-hundred micro liter of various test media was loaded into
each well of 12-
well plates prior to the addition of cells. Adherent pluripotent cells were
dissociated with
TrypLETm (Invitrogen) for 5 minutes or until fully detached from the culture
plates. TrypLETm
was neutralized by adding an equal volume of media to the culture. The cells
were counted,
washed, and resuspended in fresh media at a concentration of 300,000 to
1,000,000 cells/ml.
Approximately 100 .1 of this cell solution was added into each well of the 12-
well plates and the
cells were incubated at 37 C with 5% 02 and 10% CO2. Cells were again
dissociated at various
time points using 0.4 ml TrypLETm, which was subsequently neutralized with
equal volumes of
10% FBS in DMEM. The cells were counted by flow cytometry. 5000 count bright
beads were
added to each sample as internal control (approximately 200 beads were counted
for each
sample). All experiments were performed in triplicates.
[00075] Example 2: Growth factors for survival and short term growth.
[00076] TeSR medium contains six growth factors, in addition to those
present in the basal
medium, fibroblast growth factor (FGF), transforming growth factor beta (TGF-
f3), y-
aminobutyric acid (GABA), pipecolic acid, lithium chloride (LiC1), and insulin
(Table 1). A
basic nutrient medium (NM) was created containing all TeSRTm components with
the exception
of these six growth factors. About 2 x 105 H1 ES cells were dissociated and
plated on
MatrigelTM. The survival index was determined after 24 h. NM alone could not
support cell
survival after dissociation. The addition of insulin to NM resulted in cell
survival similar to that
observed with TeSRTm, but did not support cell growth (FIG. 1A). The addition
of both insulin
(2Oug/m1) and FGF2 (10Ong/m1) supported cell survival and additionally led to
cell growth in 96
h that was comparable to that observed using TeSRTm medium (FIG. 1B). Thus, NM

supplemented with FGF and Insulin supports human ES cell culture. Twelve
different basal
nutrient media supplemented as described above were able to support cell
survival and growth
(FIG. 1E).
[00077] Example 3: L-ascorbic acid supports short-term proliferation.
[00078] NM contains 11 nutritional components, i.e., DMEM/F12, trace
elements B, trace
elements C, L-ascorbic acid, thiamine, selenium, L-glutamine, BSA, BME, sodium
bicarbonate
(NaHCO3), and transferrin (Table 1). DMEM/F12 serves as basal medium and
NaHCO3 is used
-13-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
to modify the pH. To determine which other nutritional components were
essential when insulin
and FGF were present, each factor was added individually to DMEM/F12, NaHCO3,
insulin, and
FGF. None of the nutritional factors were essential for survival after
passaging, but L-ascorbic
acid (64mg/L) was necessary for cell proliferation after passaging (FIG. 1C).
L-ascorbic acid,
known as Vitamin C, is a major antioxidant and cofactor of several enzymes.
Hydroxyproline
could partially substitute for L-ascorbic acid. Human ES cells plated in
DMEM/F12, NaHCO3,
L-ascorbic acid, insulin, and FGF (Defined Factors 5, "DF5," Table 1)
maintained similar
morphology as human ES cells plated into TeSR.
[00079] Example 4: Media components for extended passage.
[00080] DF5 supported cell growth for only one passage. After the second
passage, cells
attached poorly and eventually died (FIG. 1C). Cells could be passaged in
NM+Insulin+FGF
(data not shown) and DF12 (Fig. 1D, Table 1), suggesting that one or more
factor present in
NM+Insulin+FGF and DF12 is important for extended passage. Each nutritional
factor present
in NM was added individually to DF5 to determine its ability to support cell
expansion after
multiple passages. Addition of selenium alone was sufficient to support cell
proliferation
through multiple passages (FIG. 1D, DF5+Selenium, "DF5S," Table 1).
[00081] DF5S was used to expand H1 cells. Cells grown in DF5S were more
prone to
differentiate than cells grown in TeSRTm. However, H1 cells could be grown for
several weeks
(more than 15 passages), during which the cells maintained human ES cell
morphology and high
levels of OCT4 expression (FIG. 1E, FIG. 1F). H1 cells grown in DF5S to which
either NODAL
(10Ong/m1) or TGF- 0 (2ng/m1) was added expressed significantly higher levels
of NANOG
mRNA, compared to H1 cells cultured in DF5S. DF5S +NODAL also supported
pluripotency of
the two tested human iPS cell lines, as determined by high expression of the
pluripotency marker
OCT4. All cells (hES cells and iPS cells) grown in DF5S with either NODAL or
TGF- 0
maintained a normal karyotype after long-term passage.
[00082] Example 5: Hypoxia improves cell growth and cloning.
[00083] H1 cells grew faster in the DF5S medium compared to cells grown in
TeSRTm
(Figure 1C and 2A). To optimize pluripotent cell growth conditions, cells were
grown in DF5S
with varying osmolarity, pH, oxygen level, and CO2 level. To increase assay
sensitivity only
5,000 cells were seeded in each well and analyzed for survival (24 h) and
proliferation (124 h).
The greatest improvements were noted when 02 and CO2 levels were varied.
Ordinary culture
-14-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
conditions use oxygen at ¨15% and CO2 at 5% (015C5). Higher CO2 often led to
slightly
higher survival after 24 hours. Lower oxygen levels increased cell growth in
both DF5S and
TeSRTm. Oxygen at 15% with CO2 at 10% (015C10), and oxygen at 5% with CO2 at
10%
(05C10) increased cell survival (FIG. 2A). Cells failed to thrive at higher 02
levels (015C5 and
015C10), while they proliferated at lower oxygen levels (05C10) (FIG. 2A).
Cells in DF5S
grew faster than those grown in TeSRTm, and grew fastest at 5% 02 and 10% CO2
(FIG. 2A).
Further decreases in oxygen level to 2% reduced cell growth compared to 5% 02.
[00084] To determine cloning efficiency at various oxygen and CO2
concentrations, 500
cells were seeded into each well. Even at low oxygen, cloning efficiency was
too low (<2%) to
determine effects of various conditions on cloning. HA100, a ROCK inhibitor
known to increase
cloning efficiency, was used to increase cloning efficiency for testing oxygen
and CO2
concentrations. Conditioned medium (CM), known to be the best medium for
cloning, was used
as control. The addition of HA100 significantly improved cloning efficiency in
CM at both
05C10 and 015C5 and cloning efficiency was higher at the 05C10 than 015C5
(FIG. 2B).
Cloning efficiency of cells in DF5S was comparable to that of cells in CM
under both conditions
(FIG. 2B).
[00085] Because of the positive impact of hypoxia on cell survival, some
of the
subsequent examples employ hypoxic conditions when cells were maintained at
low density.
However, when cells were not cultured at low cell density, experiments were
conducted under
both normoxic and hypoxic conditions (FIG. 2B).
[00086] Example 6: Improved iPS cell cloning efficiency.
[00087] To determine how DF5S affects cloning efficiency, two iPS cell
lines were grown
in DF5S and plated at cloning density (approximately 500 cells per 12-well
plate well) in the
presence of HA100. The cloning efficiency of iPS cells grown in DF5S was lower
than that of
iPS cells grown in either TeSRTm or CM (FIG. 2C), suggesting that a factor
that enhances
cloning efficiency is present in TeSRTm medium, but absent from DF5S. To
identify such factor,
individual TeSRTm components were added individually to DF5S and tested for
effect on cloning
efficiency. The addition of holo-transferrin to DF5S (DF5SFe) resulted in
cloning efficiency
comparable to that using TeSRTm (FIG. 2D). Transferrin also lead to noticeable
improvement of
cloning efficiency of H1 cells in DF5S medium.
-15-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
[00088] The ROCK inhibitors HA100 and Y27632, and blebbistatin in DMEM/F12
supplemented with insulin, trasferrin, selenium, L-ascorbic acid, FGFs, and
TGF- 0 (or NODAL;
"E8"), increased cloning efficiency of H1 cells (FIG. 2E), which was further
increased by the
addition of transferrin and by culture under hypoxic conditions (FIG. 2F)).
The cells maintained
a normal karyotype after more than 25 passages.
[00089] Example 7: NODAL and TGF-B support long-term maintenance of H1 and
iPS
cell pluripotency in albumin-free media.
[00090] As described in Example 3, human pluripotent cells, such as H1,
H9, and iPS
cells, could be grown and passaged over 15 times in DF5S but were prone to
differentiate, such
that extra care is needed to sustain pluripotency in DF5S. Because
pluripotency could be
maintained more easily in TeSRTm, growth factors present in TeSRTm were added
individually to
DF5SFe used to grow H1 cells that were previously cultured in DF5S without
differentiating to
identify factors supporting long-term pluripotency. Cells were passaged
approximately one day
after reaching confluency, facilitating cell differentiation, and Oct4
expression, assessed by flow
cytometry, was used as indicator of pluripotency.
[00091] Human pluripotent cells grown in DF5SFe elongated and lined up
along each
other, resembling a "spindle" shape just prior to differentiation. This
phenotype is often
observed at the onset of neural differentiation that is usually suppressed by
the TGF-13/BMP
pathway. Thus, recombinant proteins of the TGF-13 pathway were tested for
their ability to
support long-term pluripotency. DF5SFe supplemented with NODAL ("E8 (NODAL)")
used at
TeSRTm concentration sustained high Oct4 expression. DF5SFe supplemented with
TGF-13 ("E8
(TGF-13)") used at TeSRTm concentration (0.6ng/m1) supported low levels of
Oct4 expression but
was able to maintain high Oct4 expression when used at higher concentration
(lng/m1).
[00092] Human ES cell lines, such as H1 and H9 have a culture history that
includes
exposure to various complex culture components, such as FBS, feeder cells, and
knockout serum
replacer. Exposure to these components could conceivably create dependency on
these
components and, consequently, alter cellular response to simplified media.
Culture history might
play a lesser role for iPS cells, derived from reprogrammed somatic cells, as
derivation
conditions are less complex. Therefore, different factors were tested with two
original lentiviral
iPS cell lines (Yu, et at., Science 318:1917 (2007)) grown in DF5SFe. Cells
were transferred
from MEF plates directly into DF5SFe medium for one passage and then passaged
into various
-16-

CA 02807418 2015-03-25
growth factor conditions. The addition of either TGF-13 (2ng/m1) or NODAL
(10Ong/m1) to
DF5SFe ("E8 (TGF-13)" and "E8 (NODAL)," respectively) supported long-term
pluripotency of
iPS cells. Pluripotency surface markers SSEA4, SSEA3, Tra-1-60, and Tra-1-81
were also
expressed. Cells with normal karyotypes were continuously maintained for more
than 20
passages. The cells were capable of forming teratomas 5-7 weeks after
injection into severe
combined immunodeficient (SCID) mice.
[00093] E8 (TGF-13) and E8 (NODAL) supported pluripotency of every
pluripotent cell
line tested, i.e., two human ES cell lines (H1 and H9) and five iPSC lines for
more than 25
passages (approximately 3 months) with no sign of differentiation (FIG. 3). H1
ES cells grown
in E8 media have a similar gene expression profile compared to H1 ES cells
grown in TeSkrm
(FIG. 3B).
[00094] Example 8: Derivation of iPS cells in albumin-free media.
[00095] Available reprogramming protocols include incubation of the cells
in FBS in the
first several days after viral transduction or electroporation, prior to
switching the cells to
UM100 (US Patent No. 7,439,064) or CM. The simplified media described in
previous
Examples were tested for their ability to support reprogramming. ES-derived
somatic cells could
be reprogrammed efficiently in DF5S medium using lentivirus or episomal
vectors with or
without an initial 2 day culture in FBS-containing media. However, DF5S did
not support
reprogramming of primary foreskin cells using Nanog, Oct4, Sox2 and Lin28.
DF5SFe
supported reprogramming of foreskin and adult cells on Matrigel or MEFs using
improved
lentivirus (Ebert et al., Nature 457(7227):277-280 (2009)) when the cells were
initially incubated
in FBS-containing medium. While DF5SFe was as effective as CM in supporting
reprogramming, initial exposure to FBS appeared important for reprogramming.
[00096] Foreskin cells grow significantly slower in DF5SFe than in FBS
media. To
determine growth factors that can help primary foreskin cell growth,
individual growth factors
contained in FBS were tested. The FGF family of growth factors has several
members, one or
more of which is commonly used for fibroblast culture. DF5SFe contains 100
ng/ml zebrafish
recombinant FGF2. Each FGF family member was tested for its ability to support
foreskin cell
growth. Foreskin cells were aliquoted into the well of culture plates and
incubated for 24 hours
in DF5SFe minus FGF. Individual FGF types were added at 100 ng/ml for 96 h.
FGF1, zFGF2,
-17-

CA 02807418 2015-03-25
. ,
FGF4, FGF6, and FGF9 supported foreskin cell growth most effectively, but none
supported cell
growth as well as FBS-containing media (FIG. 4A). To identify if a non-FGF
family member
growth factor could promote foreskin cell growth comparable to that seen with
FBS, several
known fibroblast growth-promoting factors were tested. Hydrocortisone (FIG.
4B), its
derivatives, and dexamethasone added to DF5SFe to replace FBS improved cell
growth
significantly. DF5SFe+hydrocortisone ("DF5SFeC") also improved iPS cell
cloning efficiency.
[00097] To determine if DF5S-based media can be used for viral-free iPS
cell derivation,
foreskin cells were reprogrammed using a viral-free episomal vector, as
described in Yu et al.,
Science 324:797 (2009), at hypoxic conditions (05C10). Plasmid combinations #4

(pEP4EP2SCK2MEN2L and pEP4E02SET2K, Table 3), #6 (pEP4E02SEN2L,
pEP4E02SET2K and pEP4E02SEM2K, Table 3), and #19 (pEP4E02SEN2K,
pEP4E02SET2K, and pCEP4-M2L, Table 3) were used, and 2 clones were isolated
from 106
cells after secondary passage.
[00098] Table 3 Reprogramming vector components and vector combinations
SEQ ID
Component Abbr. Source Accession # or
sequence
NO
OCT4 0 hESC 1 NM 002701
SOX2 S hESC 2 NM 003106
NANOG N hESC 3 NM 024865
LIN28 L hESC 4 NM 024674
c-Myc M hESC 5 NM 002467
KLF4 K hESC 6 NM 004235
SV40 T T pBABE-puro SV40 LT p 7 EF579667
TERT TERT pBABE-hygro-hTERT 8 NM 198253
IRES2 2 pIRES2EGFP 9
CMV C 10
EFla E 11
Vector Combinations
Combination
Plasmids Components
Number
-18-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
pEP4EP2SC pEP4-EF1a-OCT4-IRES2-S0X2-CMV-KLF4-IRES2-c-Myc-
K2MEN2L EFla-NANOG-IRES2-LN28
4
pEP4E02SE
pEP4-EF1a-OCT4-IRES2-S0X2-EF1a-SV40T-IRES2-KLF4
T2K
pEP4E02SE
pEP4-EF1a-OCT4-IRES2-S0X2-EF1a-NANOG-IRES2- LN28
N2L
pEP4E02SE
6 pEP4-EF1a-OCT4-IRES2-S0X2-EF1a-SV40T-IRES2-KLF4
T2K
pEP4E02SE
pEP4-EF1a-OCT4-IRES2-S0X2-EF1a-c-Myc-IRES2-KLF4
M2K
pEP4E02SE
pEP4-EF1a-OCT4-IRES2-S0X2-EF1a-NANOG-IRES2-KLF4
N2K
19 pEP4E02SE
pEP4-EF1a-OCT4-IRES2-S0X2-EF1a-SV40T-IRES2-KLF4
T2K
pCEP4-M2L pCEP4-CMV-c-Myc-IRES2-LN28
[00099] Plasmid combinations #6 and #19 were used for the reprogramming.
In order to
enhance the plasmid entry into the nucleus, ENBA mRNA was electroporated along
with
plasmid DNA. Around one million cells were transferred onto two 6-well plates
in DF5SFeC for
days. Medium was then switched to DF5SFe for another 18-25 days. Cells of some
of the
wells were passaged for a second time using a 1:6 ratio at different time
points. Plasmid
combination #19 generated more colonies than plasmid combination #6, but most
of them did
not resemble typical human ES cell morphology. After approximately 25 days,
human ES cell-
like colonies appeared on the primary plate for both plasmid combinations,
with an estimated 24
reprogrammed cells per million foreskin cells using plasmid combination #19
and 8
reprogrammed cells per million foreskin cells using plasmid combination #6.
The number of
human ES cell-like colonies significantly increased after the secondary
passage plates, with an
estimated >500/million foreskin cells for each plasmid combination. The
increase in the iPS cell
colonies on secondary passage plates are likely due to the split of iPS cells
on the primary plates.
In some instances, primary plates did not have any colonies resembling typical
human ES cell
-19-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
morphology, but many iPS cells appeared after secondary passage, suggesting
that some iPS
cells could not be identified, possibly because they were mixed with somatic
cells.
[000100] Cells of the iPS cell colonies derived in DF5SFe started to
differentiate after only
two passages. Six iPS cell colonies were picked from the primary plate and
transferred directly
into Nodal-containing DF5SFeN (E8 (NODAL)). These cells could be maintained in
E8 (Nodal)
for more than 15 passages, maintaining their ES cell-like morphology similar
to that observed
using TeSRTm. The cells had normal karyotypes, expressed Oct4 and SSEA4 (FIG.
4C), and
formed teratomas in SCID mice 5-7 weeks after injection.
[000101] Foreskin fibroblasts were also reprogrammed in E8 medium. Global
gene
expression of iPS cells derived in E8 medium was similar to that of H1 cells
(FIG. 4D and E) or
iPS cells derived on feeder cells (FIG. 4D and F). Pluripotency markers were
highly expressed
in both ES and iPS cells, while fibroblast specific marker genes were not
expressed (FIG. 4D).
Also, iPS cell could be derived in E8 media using various strategies, e.g.,
using lentiviral or
episomal vectors.
[000102] Example 9: Derivation of iPS cells from patient cell lines in
albumin-free media.
[000103] To determine if cells from adult donors could be reprogrammed
using viral-free
episomal vectors in the simplified media, two million cells of the patient
cell lines OAT or
PRPT8 were electroporated with plasmid combinations #4 or #6, along with EBNA
mRNA, and
transferred onto two 10 cm plates. To maximize reprogramming, FBS-containing
media was
used for the first 6 days. Cells were kept at 015C5 to match regular adult
cell maintenance
conditions. Medium was then switched to DF5SFe for another 14-21 days. The
cells of one
plate were passaged at a 1:2 ratio at different time points. Plasmid
combination #6 generated
more colonies (approximately 5 per one million cells) than #4, but most of the
cells did not
resemble typical human ES cell morphology. After approximately 22 days, human
ES cell-like
colonies appeared on the primary plate for plasmid combination #4. Many more
human ES cell-
like colonies appeared on the secondary passage plates when plasmid
combination #6 was used,
with an estimate of approximately 40 colonies per million cells. No iPS cells
were produced
when using plasmid combination #4. The iPS cell colonies emerged in the middle
of other
densely-populated cells on the primary plate and could not grow beyond their
boundary.
However, colonies on the secondary plates expanded to large sizes suitable for
colony isolation.
Colonies were picked and directly transferred into TeSRTm, and 32 picked
colonies survived and
-20-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
displayed ES-cell morphology. Genetic analysis confirmed that these colonies
were derived
from the OAT cell line and exhibited a normal karyotype.
[000104] To improve adult cell reprogramming efficiency, TGF-I3 was added
to the
reprogramming media. iPS clones were not increased significantly, however, the
total number of
colonies increased significantly. When TGF-I3 was removed from the media at
the time of
hydrocortisone removal, the number of iPS cell colonies increased
significantly, suggesting that
TGF-B supports reprogramming in the first few days of the process.
[000105] Many seemingly non-iPS clones can generate iPS clones after
secondary passage,
suggesting that iPS cell derivation might be inhibited by surrounding cells.
Several reagents
were tested for their ability to overcome this effect. Butyrate improved
reprogramming
efficiency. An approximately 10-fold increase in reprogramming efficiency of
foreskin cells was
observed when both TGF-I3 and butyrate were added to the media (FIG. 5B). TGF-
I3 appeared to
exhibit its positive effects during early stages of reprogramming, while
butyrate had a positive
role in the later stage. TGF-I3 addition led to increased numbers of colonies
during
reprogramming, but the number of true iPS cell colonies remained low. Butyrate
did not
increase the number of colonies, but improved the ratio of true iPS cell to
non-iPS cell colonies
significantly (FIG. 5C).
[000106] Using TGF-I3 and butyrate enabled successful reprogramming of
somatic cells
from an adult individual under completely defined conditions using the
episomal vector system.
iPS cells were derived from three independent adult somatic cell lines (OAT,
GRC M1-29, and
PRPF8-2) at an efficiency of 1-100 out of 1 x 106 PRPF8-2 cells and 1 out of
100,000 cells
(GRC 1-29).
[000107] Example 10. Derivation of iPS cells from an adult individual in
completely
defined conditions.
[000108] A biopsy was taken from the skin of a male adult donor, washed
several times
with Hank's Buffered Salt Solution (HBSS) containing antibiotics and
antimycotic agents, and
incubated in 2 ml of 0.25% trypsin/EDTA (Table 4) or TrypLE select at 4 C
overnight. The
sample was rinsed three times, using trypsin inhibitor (Table 4) after the
second rinse. The
dermis and epidermis were separated using sterile forceps. The dermis was cut
into small pieces
and incubated in 0.75m1 enzyme solution (Table 4) with defined enzymes at room
temperature
(12-well or 24-well plate) for 3 hours. After approximately 35 minutes, tissue
structures started
-21-

CA 02807418 2013-02-01
WO 2012/019122 PCT/US2011/046796
to break down. An equal volume of medium with 1Oug/m1 polyvinylpyrrolidone
(PVP) was
added and the tissue was mechanically dissociated by pipetting up and down
about 10 times.
The sample was centrifuged at 400g for 10 minutes at room temperature and
washed twice with
fresh media/PVP. The supernatant was discarded, the pellet resuspended in 3m1
of complete
medium, and lml of the cell suspension was transferred into wells of 6-well
plates coated with
3 g/well vitronectin. The plates were incubated with 5% CO2 at 37 C and the
medium was
changed every day. Fibroblasts adhered to the plates while nonadherent cells
and debris were
removed when the medium was changed.
[000109] Table 4. Reagents and procedures for specimen digestion.
Trypsin/EDTA Enzyme solution Trypsin inhibitor
TrypZean lx (Sigma) HEPES containing RPMI 10mg/m1 Trypsin inhibitor
supplemented with 1mM (from Glycine max) in
TrypLE animal free sodium pyruvate, 1.0 Water or PBS
0.05%41.25% (Invitrogen) mg/ml Collagenase, 150
units/ml Hyaluronidase,
and 140 units/ml DNase I
(Roche)
[000110] After 20 days, reprogramming plasmids were introduced into the
fibroblasts using
electroporation. Within the next 25 days, multiple iPS colonies emerged and
were picked for
further analysis. Reprogramming efficiency was about 10 out of 1 million
electroporated
fibroblasts, without secondary passaging. iPS cells were further passaged to
isolate vector-free
cell lines.
[000111] Example 11. Derivation of iPS cells from an adult individual in
albumin-free
media without secondary passage.
[000112] Adult fibroblasts were reprogrammed in E8 (DMEM/F12 supplemented
with
insulin, transferrin, selenium, L-ascorbic acid, FGF2, and TGF- 0 (or NODAL))
following the
general protocol illustrated in FIG. 6A. Reprogrammed iPS cell lines
maintained in E8 for more
than 20 passages continued to express pluripotency markers OCT4 and SSEA4
(FIG. 6B).
[000113] E8 medium significantly enhanced reprogramming efficiency compared
to
reprogramming efficiencies using mouse fibroblast feeder cells (MEF) (FIG. 7A)
or TeSRTm
(FIG. 7B). Butyrate (100 M) further enhanced reprogramming efficiency in the
presence of
TGF-I3 (E8) or in the absence of TGF-I3 (E8 without of TGF-f3, i.e., DF5SFe )
(FIG. 7C).
-22-

CA 02807418 2015-03-25
[000114] Example 12. Cryopreservation of pluripotent stem cells in an
albumin-free media.
[000115] Pluripotent cells were cultured in 6-well plates in E8 medium,
essentially as
described above. The culture medium was aspirated from each well and the cells
were washed
twice with 1.0mL EDTA/PBS (0.5mM EDTA in PBS, osmalority 340). The cells were
then
incubated at 37 C in EDTA/PBS for 5 minutes. The PBS/EDTA was removed, and
the cells
were rinsed swiftly with lml of E8 medium. The cells were then resuspended in
an equal
volume of 20% dimethyl sulfoxide (DMSO) and E8 medium (final concentration:
10% DMSO in
E8 medium), aliquoted into cryogenic vials, and frozen at -80 C using a
CRYOBOXTM. The
cells were subsequently moved into a liquid nitrogen tank.
[000116] The invention has been described in connection with what are
presently
considered to be the most practical and preferred embodiments. However, the
present invention
has been presented by way of illustration and is not intended to be limited to
the disclosed
embodiments. The scope of the claims should not be limited by the preferred
embodiments set
forth in the examples, but should be given the broadest interpretation
consistent with the
description as a whole.
-23-

Representative Drawing

Sorry, the representative drawing for patent document number 2807418 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2017-04-04
(86) PCT Filing Date 2011-08-05
(87) PCT Publication Date 2012-02-09
(85) National Entry 2013-02-01
Examination Requested 2013-02-21
(45) Issued 2017-04-04

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $263.14 was received on 2023-07-03


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-08-05 $125.00
Next Payment if standard fee 2024-08-05 $347.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $400.00 2013-02-01
Request for Examination $800.00 2013-02-21
Maintenance Fee - Application - New Act 2 2013-08-05 $100.00 2013-07-26
Maintenance Fee - Application - New Act 3 2014-08-05 $100.00 2014-07-22
Maintenance Fee - Application - New Act 4 2015-08-05 $100.00 2015-07-10
Maintenance Fee - Application - New Act 5 2016-08-05 $200.00 2016-07-06
Final Fee $300.00 2017-02-16
Maintenance Fee - Patent - New Act 6 2017-08-07 $200.00 2017-07-05
Maintenance Fee - Patent - New Act 7 2018-08-06 $200.00 2018-07-11
Maintenance Fee - Patent - New Act 8 2019-08-06 $200.00 2019-07-10
Maintenance Fee - Patent - New Act 9 2020-08-05 $200.00 2020-07-15
Maintenance Fee - Patent - New Act 10 2021-08-05 $255.00 2021-07-14
Maintenance Fee - Patent - New Act 11 2022-08-05 $254.49 2022-06-29
Maintenance Fee - Patent - New Act 12 2023-08-07 $263.14 2023-07-03
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
WISCONSIN ALUMNI RESEARCH FOUNDATION
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Cover Page 2013-04-10 1 27
Abstract 2013-02-01 1 53
Claims 2013-02-01 4 109
Drawings 2013-02-01 17 831
Description 2013-02-01 23 1,313
Description 2015-03-25 23 1,319
Claims 2015-03-25 3 103
Claims 2016-03-10 3 111
PCT 2013-02-01 13 487
Assignment 2013-02-01 3 85
Prosecution-Amendment 2013-05-01 2 56
Prosecution-Amendment 2013-02-21 2 47
Prosecution-Amendment 2014-09-29 3 136
Prosecution-Amendment 2015-03-25 14 619
Examiner Requisition 2015-09-14 4 243
Amendment 2016-03-10 11 426
Amendment 2016-03-10 2 60
Final Fee 2017-02-16 2 56
Cover Page 2017-03-01 1 28

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :